Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Vaccine ; 41(21): 3308-3316, 2023 05 16.
Article in English | MEDLINE | ID: mdl-37085457

ABSTRACT

In the past decade, camelid nanobodies have been developed for multiple applications, including immuno-imaging, cancer immunotherapy, and antiviral therapeutics. Despite the prevalence of these approaches, nanobodies have rarely been used to assess the potency of vaccine antigen candidates, which are primarily based on mAb binding approaches. In this work, we demonstrate that a nanobody-based ELISA method is suitable for characterization of a leading respiratory syncytial virus (RSV) vaccine candidate, RSVPreF3. This nanobody, F-VHH-L66, compares similarly with AM14, an antibody well-known to be specific for the prefusion form of the RSV surface fusion glycoprotein, RSV F. ELISA assays based on F-VHH-L66 were specific for the trimeric, prefusion form of RSV F, the antigen conformation that best generates neutralizing antibodies. Additionally, the F-VHH-L66-based ELISA proved accurate, linear, and stability-indicating. Statistical analysis of 65 independent F-VHH-L66-based ELISA experiments indicated assay performance similar to that of ELISA assays based on AM14. Moreover, the binding kinetics of F-VHH-L66 to RSVPreF3 are comparable to those of AM14 when measured by surface plasmon resonance (SPR). Finally, F-VHH-L66 neutralized RSV(A) with similar efficacy as AM14; this bioactivity data further supports its use as an alternative to AM14 for pre-fusion specific structural characterization of RSVPreF3.


Subject(s)
Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus Vaccines , Respiratory Syncytial Virus, Human , Single-Domain Antibodies , Humans , Antibodies, Viral , Antibodies, Neutralizing , Antiviral Agents , Viral Fusion Proteins , Respiratory Syncytial Virus Infections/prevention & control
2.
PLoS One ; 13(9): e0198303, 2018.
Article in English | MEDLINE | ID: mdl-30192757

ABSTRACT

Dengue virus (DENV), an arthropod-borne ("arbovirus") virus, causes a range of human maladies ranging from self-limiting dengue fever to the life-threatening dengue shock syndrome and proliferates well in two different taxa of the Animal Kingdom, mosquitoes and primates. Mosquitoes and primates show taxonomic group-specific intolerance to certain codon pairs when expressing their genes by translation. This is called "codon pair bias". By necessity, dengue viruses evolved to delicately balance this fundamental difference in their open reading frames (ORFs). We have undone the evolutionarily conserved genomic balance in the DENV2 ORF sequence and specifically shifted the encoding preference away from primates. However, this recoding of DENV2 raised concerns of 'gain-of-function,' namely whether recoding could inadvertently increase fitness for replication in the arthropod vector. Using mosquito cell lines and two strains of Aedes aegypti we did not observe any increase in fitness in DENV2 variants codon pair deoptimized for humans. This ability to disrupt and control DENV2's host preference has great promise towards developing the next generation of synthetic vaccines not only for DENV but for other emerging arboviral pathogens such as chikungunya virus and Zika virus.


Subject(s)
Dengue Virus/genetics , Genome, Viral , Virus Replication/genetics , Aedes/virology , Animals , Cell Line , Chlorocebus aethiops , Vero Cells
3.
Antiviral Res ; 150: 217-225, 2018 02.
Article in English | MEDLINE | ID: mdl-29288700

ABSTRACT

Many flaviviruses, such as Zika virus (ZIKV), Dengue virus (DENV1-4) and yellow fever virus (YFV), are significant human pathogens. Infection with ZIKV, an emerging mosquito-borne flavivirus, is associated with increased risk of microcephaly in newborns and Guillain-Barré syndrome and other complications in adults. Currently, specific therapy does not exist for any flavivirus infections. In this study, we found that erythrosin B, an FDA-approved food additive, is a potent inhibitor for flaviviruses, including ZIKV and DENV2. Erythrosin B was found to inhibit the DENV2 and ZIKV NS2B-NS3 proteases with IC50 in low micromolar range, via a non-competitive mechanism. Erythrosin B can significantly reduce titers of representative flaviviruses, DENV2, ZIKV, YFV, JEV, and WNV, with micromolar potency and with excellent cytotoxicity profile. Erythrosin B can also inhibit ZIKV replication in ZIKV-relevant human placental and neural progenitor cells. As a pregnancy category B food additive, erythrosin B may represent a promising and easily developed therapy for management of infections by ZIKV and other flaviviruses.


Subject(s)
Antiviral Agents/pharmacology , Erythrosine/pharmacology , Flavivirus/drug effects , Flavivirus/enzymology , Protease Inhibitors/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Antiviral Agents/chemistry , Cells, Cultured , Dose-Response Relationship, Drug , Erythrosine/chemistry , Flavivirus/genetics , Flavivirus Infections/virology , Gene Expression Regulation, Viral/drug effects , Humans , Models, Molecular , Molecular Conformation , Protease Inhibitors/chemistry , Protein Binding , RNA Helicases/antagonists & inhibitors , RNA Helicases/chemistry , RNA Helicases/genetics , RNA Helicases/metabolism , Serine Endopeptidases/chemistry , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Structure-Activity Relationship , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism
4.
Cell Res ; 27(8): 1046-1064, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28685770

ABSTRACT

Recent outbreaks of Zika virus (ZIKV) highlight an urgent need for therapeutics. The protease complex NS2B-NS3 plays essential roles during flaviviral polyprotein processing, and thus represents an attractive drug target. Here, we developed a split luciferase complementation-based high-throughput screening assay to identify orthosteric inhibitors that directly target flavivirus NS2B-NS3 interactions. By screening a total of 2 816 approved and investigational drugs, we identified three potent candidates, temoporfin, niclosamide, and nitazoxanide, as flavivirus NS2B-NS3 interaction inhibitors with nanomolar potencies. Significantly, the most potent compound, temoporfin, not only inhibited ZIKV replication in human placental and neural progenitor cells, but also prevented ZIKV-induced viremia and mortality in mouse models. Structural docking suggests that temoporfin potentially binds NS3 pockets that hold critical NS2B residues, thus inhibiting flaviviral polyprotein processing in a non-competitive manner. As these drugs have already been approved for clinical use in other indications either in the USA or other countries, they represent promising and easily developed therapies for the management of infections by ZIKV and other flaviviruses.


Subject(s)
Molecular Docking Simulation , Viral Nonstructural Proteins , Virus Replication/physiology , Zika Virus , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Cell Line , Female , Humans , Neural Stem Cells/metabolism , Neural Stem Cells/virology , Placenta/metabolism , Placenta/virology , Pregnancy , RNA Helicases/chemistry , RNA Helicases/genetics , RNA Helicases/metabolism , Serine Endopeptidases/chemistry , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Virus Replication/drug effects , Zika Virus/chemistry , Zika Virus/physiology , Zika Virus Infection/drug therapy , Zika Virus Infection/genetics , Zika Virus Infection/metabolism
5.
PLoS Pathog ; 13(5): e1006411, 2017 May.
Article in English | MEDLINE | ID: mdl-28542603

ABSTRACT

The flavivirus genome encodes a single polyprotein precursor requiring multiple cleavages by host and viral proteases in order to produce the individual proteins that constitute an infectious virion. Previous studies have revealed that the NS2B cofactor of the viral NS2B-NS3 heterocomplex protease displays a conformational dynamic between active and inactive states. Here, we developed a conformational switch assay based on split luciferase complementation (SLC) to monitor the conformational change of NS2B and to characterize candidate allosteric inhibitors. Binding of an active-site inhibitor to the protease resulted in a conformational change of NS2B and led to significant SLC enhancement. Mutagenesis of key residues at an allosteric site abolished this induced conformational change and SLC enhancement. We also performed a virtual screen of NCI library compounds to identify allosteric inhibitors, followed by in vitro biochemical screening of the resultant candidates. Only three of these compounds, NSC135618, 260594, and 146771, significantly inhibited the protease of Dengue virus 2 (DENV2) in vitro, with IC50 values of 1.8 µM, 11.4 µM, and 4.8 µM, respectively. Among the three compounds, only NSC135618 significantly suppressed the SLC enhancement triggered by binding of active-site inhibitor in a dose-dependent manner, indicating that it inhibits the conformational change of NS2B. Results from virus titer reduction assays revealed that NSC135618 is a broad spectrum flavivirus protease inhibitor, and can significantly reduce titers of DENV2, Zika virus (ZIKV), West Nile virus (WNV), and Yellow fever virus (YFV) on A549 cells in vivo, with EC50 values in low micromolar range. In contrast, the cytotoxicity of NSC135618 is only moderate with CC50 of 48.8 µM on A549 cells. Moreover, NSC135618 inhibited ZIKV in human placental and neural progenitor cells relevant to ZIKV pathogenesis. Results from binding, kinetics, Western blot, mass spectrometry and mutagenesis experiments unambiguously demonstrated an allosteric mechanism for inhibition of the viral protease by NSC135618.


Subject(s)
Enzyme Inhibitors/pharmacology , Flavivirus/drug effects , High-Throughput Screening Assays/methods , Viral Nonstructural Proteins/chemistry , Allosteric Regulation , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemistry , Flavivirus/chemistry , Flavivirus/enzymology , Flavivirus/genetics , Kinetics , Protein Conformation , RNA Helicases/antagonists & inhibitors , RNA Helicases/chemistry , RNA Helicases/genetics , RNA Helicases/metabolism , Serine Endopeptidases/chemistry , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism
6.
Emerg Infect Dis ; 23(7): 1110-1117, 2017 07.
Article in English | MEDLINE | ID: mdl-28430564

ABSTRACT

In the Western Hemisphere, Zika virus is thought to be transmitted primarily by Aedes aegypti mosquitoes. To determine the extent to which Ae. albopictus mosquitoes from the United States are capable of transmitting Zika virus and the influence of virus dose, virus strain, and mosquito species on vector competence, we evaluated multiple doses of representative Zika virus strains in Ae. aegypti and Ae. albopictus mosquitoes. Virus preparation (fresh vs. frozen) significantly affected virus infectivity in mosquitoes. We calculated 50% infectious doses to be 6.1-7.5 log 10 PFU/mL; minimum infective dose was 4.2 log 10 PFU/mL. Ae. albopictus mosquitoes were more susceptible to infection than Ae. aegypti mosquitoes, but transmission efficiency was higher for Ae. aegypti mosquitoes, indicating a transmission barrier in Ae. albopictus mosquitoes. Results suggest that, although Zika virus transmission is relatively inefficient overall and dependent on virus strain and mosquito species, Ae. albopictus mosquitoes could become major vectors in the Americas.


Subject(s)
Aedes/virology , Insect Vectors/virology , Zika Virus Infection/transmission , Zika Virus Infection/virology , Zika Virus/classification , Animals , Chlorocebus aethiops , Host-Pathogen Interactions , Vero Cells , Viral Load , Virus Replication , Zika Virus/genetics
7.
Virology ; 498: 164-171, 2016 11.
Article in English | MEDLINE | ID: mdl-27588377

ABSTRACT

Although infections of vertebrate hosts by arthropod-borne viruses may lead to pathogenic outcomes, infections of vector mosquitoes result in persistent infections, where the virus replicates in the host without causing apparent pathological effects. It is unclear how persistent infections are established and maintained in mosquitoes. Several reports revealed the presence of flavivirus-like DNA sequences in the mosquito genome, and recent studies have shown that DNA forms of RNA viruses restrict virus replication in Drosophila, suggesting that DNA forms may have a role in developing persistent infections. Here, we sought to investigate whether arboviruses generate DNA forms following infection in mosquitoes. Our results with West Nile, Dengue, and La Crosse viruses demonstrate that DNA forms of the viral RNA genome are generated in mosquito cells; however, not the entire viral genome, but patches of viral RNA in DNA forms can be detected 24h post infection.


Subject(s)
Arboviruses/physiology , DNA, Viral/genetics , Genome, Viral , Animals , Cell Line , Cells, Cultured , Culicidae/virology , DNA, Viral/chemistry , Gene Order , Open Reading Frames , Virus Replication
8.
PLoS One ; 10(6): e0130062, 2015.
Article in English | MEDLINE | ID: mdl-26098995

ABSTRACT

The flavivirus methyltransferase (MTase) is an essential enzyme that sequentially methylates the N7 and 2'-O positions of the viral RNA cap, using S-adenosyl-L-methionine (SAM) as a methyl donor. We report here that small molecule compounds, which putatively bind to the SAM-binding site of flavivirus MTase and inhibit its function, were identified by using virtual screening. In vitro methylation experiments demonstrated significant MTase inhibition by 13 of these compounds, with the most potent compound displaying sub-micromolar inhibitory activity. The most active compounds showed broad spectrum activity against the MTase proteins of multiple flaviviruses. Two of these compounds also exhibited low cytotoxicity and effectively inhibited viral replication in cell-based assays, providing further structural insight into flavivirus MTase inhibition.


Subject(s)
Antiviral Agents/pharmacology , Flavivirus Infections/drug therapy , Flavivirus/drug effects , Methyltransferases/antagonists & inhibitors , Viral Nonstructural Proteins/antagonists & inhibitors , Binding Sites , Flavivirus/enzymology , Methylation/drug effects , Microbial Sensitivity Tests , RNA Caps/metabolism , RNA, Viral/genetics , Virus Replication/drug effects
9.
J Biol Chem ; 290(25): 15746-15757, 2015 Jun 19.
Article in English | MEDLINE | ID: mdl-25953903

ABSTRACT

Interchanging Leu-119 for Pro-119 at the tip of the ß4-ß5 loop in the first FK506 binding domain (FK1) of the FKBP51 and FKBP52 proteins, respectively, has been reported to largely reverse the inhibitory (FKBP51) or stimulatory (FKBP52) effects of these co-chaperones on the transcriptional activity of glucocorticoid and androgen receptor-protein complexes. Previous NMR relaxation studies have identified exchange line broadening, indicative of submillisecond conformational motion, throughout the ß4-ß5 loop in the FK1 domain of FKBP51, which are suppressed by the FKBP52-like L119P substitution. This substitution also attenuates exchange line broadening in the underlying ß2 and ß3a strands that is centered near a bifurcated main chain hydrogen bond interaction between these two strands. The present study demonstrates that these exchange line broadening effects arise from two distinct coupled conformational transitions, and the transition within the ß2 and ß3a strands samples a transient conformation that resembles the crystal structures of the selectively inhibited FK1 domain of FKBP51 recently reported. Although the crystal structures for their series of inhibitors were interpreted as evidence for an induced fit mechanism of association, the presence of a similar conformation being significantly populated in the unliganded FKBP51 domain is more consistent with a conformational selection binding process. The contrastingly reduced conformational plasticity of the corresponding FK1 domain of FKBP52 is consistent with the current model in which FKBP51 binds to both the apo- and hormone-bound forms of the steroid receptor to modulate its affinity for ligand, whereas FKBP52 binds selectively to the latter state.


Subject(s)
Models, Molecular , Receptors, Androgen/chemistry , Receptors, Glucocorticoid/chemistry , Tacrolimus Binding Proteins/chemistry , Amino Acid Substitution , Humans , Mutation, Missense , Nuclear Magnetic Resonance, Biomolecular , Protein Structure, Quaternary , Protein Structure, Secondary , Protein Structure, Tertiary , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Tacrolimus Binding Proteins/genetics , Tacrolimus Binding Proteins/metabolism
10.
ACS Infect Dis ; 1(8): 340-9, 2015 Aug 14.
Article in English | MEDLINE | ID: mdl-26726314

ABSTRACT

Flavivirus methyltransferase (MTase) is essential for viral replication. Here we report the identification of small molecules through virtual screening that putatively bind to the SAM-binding site of flavivirus MTase and inhibit its function. Six of these computationally predicted binders were identified to show significant MTase inhibition with low micromolar inhibitory activity. The most active compounds showed broad-spectrum activity against the MTase proteins of other flaviviruses. Two of these compounds also showed low cytotoxicity and high antiviral efficacy in cell-based assays. Competitive binding analyses indicated that the inhibitors performed their inhibitory function through competitive binding to the SAM cofactor binding site of the MTase. The crystal structure of the MTase-inhibitor complex further supports the mode of action and provides routes for their further optimization as flavivirus MTase inhibitors.

11.
Protein Sci ; 24(1): 117-28, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25352331

ABSTRACT

Methylation of flavivirus RNA is vital for its stability and translation in the infected host cell. This methylation is mediated by the flavivirus methyltransferase (MTase), which methylates the N7 and 2'-O positions of the viral RNA cap by using S-adenosyl-l-methionine (SAM) as a methyl donor. In this report, we demonstrate that SAM, in contrast to the reaction by-product S-adenosyl-l-homocysteine, which was assumed previously, is copurified with the Dengue (DNV) and West Nile virus MTases produced in Escherichia coli (E. coli). This endogenous SAM can be removed by denaturation and refolding of the MTase protein. The refolded MTase of DNV serotype 3 (DNV3) displays methylation activity comparable to native enzyme, and its crystal structure at 2.1 Å is almost identical to that of native MTase. We characterized the binding of Sinefungin (SIN), a previously described SAM-analog inhibitor of MTase function, to the native and refolded DNV3 MTase by isothermal titration calorimetry, and found that SIN binds to refolded MTase with more than 16 times the affinity of SIN binding to the MTase purified natively. Moreover, we show that SAM is also copurified with other flavivirus MTases, indicating that purification by refolding may be a generally applicable tool for studying flavivirus MTase inhibition.


Subject(s)
Dengue Virus/enzymology , Dengue/virology , Methyltransferases/metabolism , S-Adenosylhomocysteine/metabolism , S-Adenosylmethionine/metabolism , West Nile Fever/virology , West Nile virus/enzymology , Crystallography, X-Ray , Dengue Virus/chemistry , Dengue Virus/metabolism , Humans , Methyltransferases/chemistry , Methyltransferases/isolation & purification , Models, Molecular , Protein Refolding , S-Adenosylhomocysteine/isolation & purification , S-Adenosylmethionine/isolation & purification , West Nile virus/chemistry , West Nile virus/metabolism
12.
Biochem J ; 458(3): 525-36, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24405377

ABSTRACT

The extensive set of NMR doublings exhibited by the immunophilin FKBP12 (FK506-binding protein 12) arose from a slow transition to the cis-peptide configuration at Gly89 near the tip of the 80's loop, the site for numerous protein-recognition interactions for both FKBP12 and other FKBP domain proteins. The 80's loop also exhibited linebroadening, indicative of microsecond to millisecond conformational dynamics, but only in the trans-peptide state. The G89A variant shifted the trans-cis peptide equilibrium from 88:12 to 33:67, whereas a proline residue substitution induced fully the cis-peptide configuration. The 80's loop conformation in the G89P crystal structure at 1.50 Å resolution differed from wild-type FKBP12 primarily at residues 88, 89 and 90, and it closely resembled that reported for FKBP52. Structure-based chemical-shift predictions indicated that the microsecond to millisecond dynamics in the 80's loop probably arose from a concerted main chain (ψ88 and ϕ89) torsion angle transition. The indole side chain of Trp59 at the base of the active-site cleft was reoriented ~90o and the adjacent backbone was shifted in the G89P crystal structure. NOE analysis of wild-type FKBP12 demonstrated that this indole populates the perpendicular orientation at 20%. The 15N relaxation analysis was consistent with the indole reorientation occurring in the nanosecond timeframe. Recollection of the G89P crystal data at 1.20 Å resolution revealed a weaker wild-type-like orientation for the indole ring. Differences in the residues that underlie the Trp59 indole ring and altered interactions linking the 50's loop to the active site suggested that reorientation of this ring may be disfavoured in the other six members of the FKBP domain family that bear this active-site tryptophan residue.


Subject(s)
Tacrolimus Binding Protein 1A/chemistry , Catalytic Domain , Crystallography, X-Ray , Humans , Magnetic Resonance Spectroscopy , Protein Conformation
13.
Virol Sin ; 28(6): 326-36, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24242363

ABSTRACT

Many flaviviruses are significant human pathogens causing considerable disease burdens, including encephalitis and hemorrhagic fever, in the regions in which they are endemic. A paucity of treatments for flaviviral infections has driven interest in drug development targeting proteins essential to flavivirus replication, such as the viral protease. During viral replication, the flavivirus genome is translated as a single polyprotein precursor, which must be cleaved into individual proteins by a complex of the viral protease, NS3, and its cofactor, NS2B. Because this cleavage is an obligate step of the viral life-cycle, the flavivirus protease is an attractive target for antiviral drug development. In this review, we will survey recent drug development studies targeting the NS3 active site, as well as studies targeting an NS2B/NS3 interaction site determined from flavivirus protease crystal structures.


Subject(s)
Antiviral Agents/isolation & purification , Drug Discovery/methods , Flavivirus/drug effects , Flavivirus/enzymology , Protease Inhibitors/isolation & purification , Viral Nonstructural Proteins/antagonists & inhibitors , Antiviral Agents/pharmacology , Drug Discovery/trends , Humans , Protease Inhibitors/pharmacology , RNA Helicases/antagonists & inhibitors , Serine Endopeptidases
14.
PLoS One ; 8(10): e76900, 2013.
Article in English | MEDLINE | ID: mdl-24130807

ABSTRACT

The methyltransferase enzyme (MTase), which catalyzes the transfer of a methyl group from S-adenosyl-methionine (AdoMet) to viral RNA, and generates S-adenosyl-homocysteine (AdoHcy) as a by-product, is essential for the life cycle of many significant human pathogen flaviviruses. Here we investigated inhibition of the flavivirus MTase by several AdoHcy-derivatives. Unexpectedly we found that AdoHcy itself barely inhibits the flavivirus MTase activities, even at high concentrations. AdoHcy was also shown to not inhibit virus growth in cell-culture. Binding studies confirmed that AdoHcy has a much lower binding affinity for the MTase than either the AdoMet co-factor, or the natural AdoMet analog inhibitor sinefungin (SIN). While AdoMet is a positively charged molecule, SIN is similar to AdoHcy in being uncharged, and only has an additional amine group that can make extra electrostatic contacts with the MTase. Molecular Mechanics Poisson-Boltzmann Sovation Area analysis on AdoHcy and SIN binding to the MTase suggests that the stronger binding of SIN may not be directly due to interactions of this amine group, but due to distributed differences in SIN binding resulting from its presence. The results suggest that better MTase inhibitors could be designed by using SIN as a scaffold rather than AdoHcy.


Subject(s)
Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Flavivirus/enzymology , Methyltransferases/antagonists & inhibitors , Methyltransferases/metabolism , S-Adenosylhomocysteine/metabolism , S-Adenosylhomocysteine/pharmacology , Adenosine/analogs & derivatives , Adenosine/pharmacology , Cell Line , Enzyme Inhibitors/adverse effects , Flavivirus/drug effects , Humans , Models, Molecular , Protein Binding , Protein Conformation , S-Adenosylhomocysteine/adverse effects , West Nile virus/drug effects , West Nile virus/enzymology
15.
PLoS Pathog ; 9(12): e1003837, 2013.
Article in English | MEDLINE | ID: mdl-24385909

ABSTRACT

T cell responses to enteric bacteria are important in inflammatory bowel disease. I2, encoded by the pfiT gene of Pseudomonas fluorescens, is a T-cell superantigen associated with human Crohn's disease. Here we report the crystal structure of pfiT at 1.7Å resolution and provide a functional analysis of the interaction of pfiT and its homolog, PA2885, with human class II MHC. Both pfiT and PA2885 bound to mammalian cells and stimulated the proliferation of human lymphocytes. This binding was greatly inhibited by anti-class II MHC HLA-DR antibodies, and to a lesser extent, by anti HLA-DQ and DP antibodies, indicating that the binding was class II MHC-specific. GST-pfiT efficiently precipitated both endogenous and in vitro purified recombinant HLA-DR1 molecules, indicating that pfiT directly interacted with HLA-DR1. Competition studies revealed that pfiT and the superantigen Mycoplasma arthritidis mitogen (MAM) competed for binding to HLA-DR, indicating that their binding sites overlap. Structural analyses established that pfiT belongs to the TetR-family of DNA-binding transcription regulators. The distinct structure of pfiT indicates that it represents a new family of T cell superantigens.


Subject(s)
Crohn Disease/immunology , Pseudomonas fluorescens/immunology , Superantigens/chemistry , Amino Acid Sequence , Animals , Binding Sites , Cells, Cultured , Crystallography, X-Ray , HLA-DR1 Antigen/metabolism , Humans , Mice , Models, Molecular , Molecular Sequence Data , Pseudomonas aeruginosa/immunology , Sequence Homology , Superantigens/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
16.
J Virol ; 86(22): 12129-37, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22933285

ABSTRACT

Retrovirus infection starts with the binding of envelope glycoproteins to host cell receptors. Subsequently, conformational changes in the glycoproteins trigger fusion of the viral and cellular membranes. Some retroviruses, such as avian sarcoma/leukosis virus (ASLV), employ a two-step mechanism in which receptor binding precedes low-pH activation and fusion. We used cryo-electron tomography to study virion/receptor/liposome complexes that simulate the interactions of ASLV virions with cells. Binding the soluble receptor at neutral pH resulted in virions capable of binding liposomes tightly enough to alter their curvature. At virion-liposome interfaces, the glycoproteins are ∼3-fold more concentrated than elsewhere in the viral envelope, indicating specific recruitment to these sites. Subtomogram averaging showed that the oblate globular domain in the prehairpin intermediate (presumably the receptor-binding domain) is connected to both the target and the viral membrane by 2.5-nm-long stalks and is partially disordered, compared with its native conformation. Upon lowering the pH, fusion took place. Fusion is a stochastic process that, once initiated, must be rapid, as only final (postfusion) products were observed. These fusion products showed glycoprotein spikes on their surface, with their interiors occupied by patches of dense material but without capsids, implying their disassembly. In addition, some of the products presented a density layer underlying and resolved from the viral membrane, which may represent detachment of the matrix protein to facilitate the fusion process.


Subject(s)
Alpharetrovirus/metabolism , Electron Microscope Tomography/methods , Membrane Fusion , Retroviridae/metabolism , Animals , Cell Line , Chickens , Computer Simulation , Cryoelectron Microscopy/methods , Fibroblasts/virology , Fluorescence Resonance Energy Transfer/methods , Glycoproteins/chemistry , Hydrogen-Ion Concentration , Image Processing, Computer-Assisted , Liposomes/chemistry , Protein Binding , Viral Envelope Proteins/chemistry
17.
J Virol ; 86(1): 364-72, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22031933

ABSTRACT

Cellular entry of Ebola virus (EBOV), a deadly hemorrhagic fever virus, is mediated by the viral glycoprotein (GP). The receptor-binding subunit of GP must be cleaved (by endosomal cathepsins) in order for entry and infection to proceed. Cleavage appears to proceed through 50-kDa and 20-kDa intermediates, ultimately generating a key 19-kDa core. How 19-kDa GP is subsequently triggered to bind membranes and induce fusion remains a mystery. Here we show that 50-kDa GP cannot be triggered to bind to liposomes in response to elevated temperature but that 20-kDa and 19-kDa GP can. Importantly, 19-kDa GP can be triggered at temperatures ∼10°C lower than 20-kDa GP, suggesting that it is the most fusion ready form. Triggering by heat (or urea) occurs only at pH 5, not pH 7.5, and involves the fusion loop, as a fusion loop mutant is defective in liposome binding. We further show that mild reduction (preferentially at low pH) triggers 19-kDa GP to bind to liposomes, with the wild-type protein being triggered to a greater extent than the fusion loop mutant. Moreover, mild reduction inactivates pseudovirion infection, suggesting that reduction can also trigger 19-kDa GP on virus particles. Our results support the hypothesis that priming of EBOV GP, specifically to the 19-kDa core, potentiates GP to undergo subsequent fusion-relevant conformational changes. Our findings also indicate that low pH and an additional endosomal factor (possibly reduction or possibly a process mimicked by reduction) act as fusion triggers.


Subject(s)
Cathepsin L/metabolism , Ebolavirus/metabolism , Hemorrhagic Fever, Ebola/enzymology , Membrane Fusion , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Cell Line , Ebolavirus/chemistry , Ebolavirus/genetics , Endosomes/enzymology , Hemorrhagic Fever, Ebola/virology , Humans , Protein Conformation , Protein Processing, Post-Translational , Viral Envelope Proteins/genetics
18.
J Virol ; 83(7): 2883-91, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19144707

ABSTRACT

Entry of ebolavirus (EBOV) into cells is mediated by its glycoprotein (GP(1,2)), a class I fusion protein whose structure was recently determined (J. E. Lee et al., Nature 454:177-182, 2008). Here we confirmed two major predictions of the structural analysis, namely, the residues in GP(1) and GP(2) that remain after GP(1,2) is proteolytically primed by endosomal cathepsins for fusion and residues in GP(1) that are critical for binding to host cells. Mass spectroscopic analysis indicated that primed GP(1,2) contains residues 33 to 190 of GP(1) and all residues of GP(2). The location of the receptor binding site was determined by a two-pronged approach. We identified a small receptor binding region (RBR), residues 90 to 149 of GP(1), by comparing the cell binding abilities of four RBR proteins produced in high yield. We characterized the binding properties of the optimal RBR (containing GP(1) residues 57 to 149) and then conducted a mutational analysis to identify critical binding residues. Substitutions at four lysines (K95, K114, K115, and K140) decreased binding and the ability of RBR proteins to inhibit GP(1,2)-mediated infection. K114, K115, and K140 lie in a small region modeled to be located on the top surface of the chalice following proteolytic priming; K95 lies deeper in the chalice bowl. Combined with those of Lee et al., our findings provide structural insight into how GP(1,2) is primed for fusion and define the core of the EBOV RBR (residues 90 to 149 of GP(1)) as a highly conserved region containing a two-stranded beta-sheet, the two intra-GP(1) disulfide bonds, and four critical Lys residues.


Subject(s)
Ebolavirus/physiology , Glycoproteins/chemistry , Viral Fusion Proteins/chemistry , Virus Internalization , Amino Acid Sequence , Amino Acid Substitution/genetics , Binding Sites , Cell Line , DNA Mutational Analysis , Ebolavirus/genetics , Glycoproteins/genetics , Glycoproteins/metabolism , Humans , Mass Spectrometry , Models, Molecular , Molecular Sequence Data , Viral Fusion Proteins/genetics , Viral Fusion Proteins/metabolism
19.
Crit Rev Biochem Mol Biol ; 43(3): 189-219, 2008.
Article in English | MEDLINE | ID: mdl-18568847

ABSTRACT

Recent work has identified three distinct classes of viral membrane fusion proteins based on structural criteria. In addition, there are at least four distinct mechanisms by which viral fusion proteins can be triggered to undergo fusion-inducing conformational changes. Viral fusion proteins also contain different types of fusion peptides and vary in their reliance on accessory proteins. These differing features combine to yield a rich diversity of fusion proteins. Yet despite this staggering diversity, all characterized viral fusion proteins convert from a fusion-competent state (dimers or trimers, depending on the class) to a membrane-embedded homotrimeric prehairpin, and then to a trimer-of-hairpins that brings the fusion peptide, attached to the target membrane, and the transmembrane domain, attached to the viral membrane, into close proximity thereby facilitating the union of viral and target membranes. During these conformational conversions, the fusion proteins induce membranes to progress through stages of close apposition, hemifusion, and then the formation of small, and finally large, fusion pores. Clearly, highly divergent proteins have converged on the same overall strategy to mediate fusion, an essential step in the life cycle of every enveloped virus.


Subject(s)
Viral Fusion Proteins/chemistry , Viral Fusion Proteins/metabolism , Virus Internalization , Animals , Cell Membrane/metabolism , Humans , Hydrogen-Ion Concentration , Receptors, Virus/chemistry , Receptors, Virus/metabolism , Viral Fusion Proteins/genetics
20.
J Virol ; 82(6): 3131-4, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18184714

ABSTRACT

We previously showed that the cysteines flanking the internal fusion peptide of the avian sarcoma/leukosis virus subtype A (ASLV-A) Env (EnvA) are important for infectivity and cell-cell fusion. Here we define the stage of fusion at which the cysteines are required. The flanking cysteines are dispensable for receptor-triggered membrane association but are required for the lipid mixing step of fusion, which, interestingly, displays a high pH onset and a biphasic profile. Second-site mutations that partially restore infection partially restore lipid mixing. These findings indicate that the cysteines flanking the internal fusion peptide of EnvA (and perhaps by analogy Ebola virus glycoprotein) are important for the foldback stage of the conformational changes that lead to membrane merger.


Subject(s)
Alpharetrovirus/metabolism , Cysteine/physiology , Glycoproteins/physiology , Membrane Fusion , Viral Proteins/physiology , Cysteine/chemistry , Cysteine/metabolism , Glycoproteins/metabolism , Viral Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...